1
|
Wang Y, Li S. Continuous synthesis of PEGylated MIL-101(Cr) nanoparticles for neuroprotection. RSC Adv 2025; 15:12020-12027. [PMID: 40242494 PMCID: PMC12002162 DOI: 10.1039/d4ra09107h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/05/2025] [Indexed: 04/18/2025] Open
Abstract
The application of metal organic frameworks (MOFs) in targeted drug delivery for ischemic stroke therapy has emerged as a hot issue recently. Although significant progress has been made in immobilizing neuroprotective agents on MOFs, environmentally friendly large-scale preparation of nano-drug-loaded MOFs with controlled size, morphology, purity and therapeutic effect remains challenging. PEGylation of MIL-101(Cr) nanoparticles with dual ligands that have the 2,2-dimethylthiazolidine (DMTD) structure was developed in this work to mitigate nervous system injury induced by ischemia/reperfusion (IR) during a stroke. A green ultrasound-assisted continuous-flow system was established for efficient production of the versatile MOF nanoparticles. Unified nanoparticles (diameter: ∼250-350 nm) were obtained with both high quality and high space-time yield (5664 kg m-3 d-1). The MOF exhibited protective activity in SH-SY5Y cells against oxygen and glucose deprivation and H2O2 insults, and prevented reactive oxygen species accumulation. The cellular uptake of the PEGylated MOFs by brain capillary endothelial cells was investigated, showing targeting capability in vitro, which proposes the biomaterial as a promising therapeutic candidate for reducing IR-induced nervous system injury.
Collapse
Affiliation(s)
- Yuheng Wang
- College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Shuirong Li
- College of Energy, Xiamen University Xiamen 361102 China
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM) Xiamen 361005 China
| |
Collapse
|
2
|
Zhong R, He H, Wang X. Novel neutrophil targeting platforms in treating Glioblastoma: Latest evidence and therapeutic approaches. Int Immunopharmacol 2025; 150:114173. [PMID: 39938169 DOI: 10.1016/j.intimp.2025.114173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/14/2025]
Abstract
Glioblastoma (GBM) is the most aggressive and lethal type of primary brain tumor, characterized by its rapid growth, resistance to conventional therapies, and a highly immunosuppressive tumor microenvironment (TME). Recent studies have highlighted the critical role of neutrophils in the progression of GBM, where they contribute to tumor growth, invasion, and treatment resistance. As a result, neutrophils have emerged as a promising target for therapeutic intervention in GBM. Various strategies are being investigated to specifically target neutrophils within the GBM environment, including using small molecules, antibodies, and nanoparticle-based methods. These approaches aim to regulate neutrophils' recruitment, activation, and functions. This study reviews the latest findings regarding the involvement of neutrophils in GBM, explores potential techniques targeting neutrophils for therapeutic purposes, and discusses current clinical studies and prospects in this rapidly evolving field. By studying the diverse functions of neutrophils in GBM, these innovative therapeutic strategies can help address some of the most significant challenges in treating this malignancy.
Collapse
Affiliation(s)
- Rui Zhong
- Department of Neurosurgery, The First People's Hospital of Lin'an District, Hangzhou 311300, China
| | - Hongmei He
- Department of Neurosurgery, The First People's Hospital of Lin'an District, Hangzhou 311300, China
| | - Xiande Wang
- Department of Neurosurgery, The First People's Hospital of Lin'an District, Hangzhou 311300, China.
| |
Collapse
|
3
|
Li H, Shan W, Zhao X, Sun W. Neutrophils: Linking Inflammation to Thrombosis and Unlocking New Treatment Horizons. Int J Mol Sci 2025; 26:1965. [PMID: 40076593 PMCID: PMC11901051 DOI: 10.3390/ijms26051965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Neutrophils play a key role in inflammatory responses and thrombosis, but their complex interactions in disease pathogenesis are not fully understood. This review examines the multifaceted roles of neutrophils, focusing on their activation, cytokine release, and formation of neutrophil extracellular traps (NETs), which contribute to host defense and thrombosis. We discuss the interaction between inflammation and coagulation, the direct effect of neutrophils on thrombus stability, and their involvement in pathological thrombotic diseases. The therapeutic potential of neutrophil drug loading in the treatment of thrombosis, as well as the clinical implications and future research directions, are highlighted. The aim of this review is to gain insight into the critical neutrophil-inflammation-thrombus axis and its potential as a therapeutic target for thrombotic diseases and to suggest possible directions for neutrophil-loaded drug therapy for thrombosis.
Collapse
Affiliation(s)
| | | | | | - Wei Sun
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.L.); (W.S.); (X.Z.)
| |
Collapse
|
4
|
Wang S, Yang L, He W, Zheng M, Zou Y. Cell Membrane Camouflaged Biomimetic Nanoparticles as a Versatile Platform for Brain Diseases Treatment. SMALL METHODS 2025; 9:e2400096. [PMID: 38461538 DOI: 10.1002/smtd.202400096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/27/2024] [Indexed: 03/12/2024]
Abstract
Although there are various advancements in biomedical in the past few decades, there are still challenges in the treatment of brain diseases. The main difficulties are the inability to deliver a therapeutic dose of the drug to the brain through the blood-brain barrier (BBB) and the serious side effects of the drug. Thus, it is essential to select biocompatible drug carriers and novel therapeutic tools to better enhance the effect of brain disease treatment. In recent years, biomimetic nanoparticles (BNPs) based on natural cell membranes, which have excellent biocompatibility and low immunogenicity, are widely used in the treatment of brain diseases to enable the drug to successfully cross the BBB and target brain lesions. BNPs can prolong the circulation time in vivo, are more conducive to drug aggregation in brain lesions. Cell membranes (CMs) from cancer cells (CCs), red blood cells (RBCs), white blood cells (WBCs), and so on are used as biomimetic coatings for nanoparticles (NPs) to achieve the ability to target, evade clearance, or stimulate the immune system. This review summarizes the application of different cell sources as BNPs coatings in the treatment of brain diseases and discusses the possibilities and challenges of clinical translation.
Collapse
Affiliation(s)
- Shiyu Wang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Longfei Yang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Wenya He
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meng Zheng
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yan Zou
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| |
Collapse
|
5
|
Fan CH, Yeh CK. Theranostic nanomaterials for intervention of the blood–brain barrier. THERANOSTICS NANOMATERIALS IN DRUG DELIVERY 2025:395-410. [DOI: 10.1016/b978-0-443-22044-9.00014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
6
|
Ranjan R, Kumar D, Singh MR, Singh D. Novel drug delivery systems in cerebral vascular disorders, transient ischaemic attack, and stroke interventions. NOVEL DRUG DELIVERY SYSTEMS IN THE MANAGEMENT OF CNS DISORDERS 2025:295-311. [DOI: 10.1016/b978-0-443-13474-6.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
Bezze A, Mattioda C, Ciardelli G, Mattu C. Harnessing cells to improve transport of nanomedicines. Eur J Pharm Biopharm 2024; 203:114446. [PMID: 39122052 DOI: 10.1016/j.ejpb.2024.114446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Efficient tumour treatment is hampered by the poor selectivity of anticancer drugs, resulting in scarce tumour accumulation and undesired off-target effects. Nano-sized drug-delivery systems in the form of nanoparticles (NPs) have been proposed to improve drug distribution to solid tumours, by virtue of their ability of passive and active tumour targeting. Despite these advantages, literature studies indicated that less than 1% of the administered NPs can successfully reach the tumour mass, highlighting the necessity for more efficient drug transporters in cancer treatment. Living cells, such as blood cells, circulating immune cells, platelets, and stem cells, are often found as an infiltrating component in most solid tumours, because of their ability to naturally circumvent immune recognition, bypass biological barriers, and reach inaccessible tissues through innate tropism and active motility. Therefore, the tumour-homing ability of these cells can be harnessed to design living cell carriers able to improve the transport of drugs and NPs to tumours. Albeit promising, this approach is still in its beginnings and suffers from difficult scalability, high cost, and poor reproducibility. In this review, we present an overview of the most common cell transporters of drugs and NPs, and we discuss how different cell types interact with biological barriers to deliver cargoes of various natures to tumours. Finally, we analyse the different techniques used to load drugs or NPs in living cells and discuss their advantages and disadvantages.
Collapse
Affiliation(s)
- Andrea Bezze
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Carlotta Mattioda
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Gianluca Ciardelli
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Clara Mattu
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy.
| |
Collapse
|
8
|
López-Espinosa J, Park P, Holcomb M, Godin B, Villapol S. Nanotechnology-driven therapies for neurodegenerative diseases: a comprehensive review. Ther Deliv 2024; 15:997-1024. [PMID: 39297726 PMCID: PMC11583628 DOI: 10.1080/20415990.2024.2401307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 11/22/2024] Open
Abstract
Neurological diseases, characterized by neuroinflammation and neurodegeneration, impose a significant global burden, contributing to substantial morbidity, disability and mortality. A common feature of these disorders, including stroke, traumatic brain injury and Alzheimer's disease, is the impairment of the blood-brain barrier (BBB), a critical structure for maintaining brain homeostasis. The compromised BBB in neurodegenerative conditions poses a significant challenge for effective treatment, as it allows harmful substances to accumulate in the brain. Nanomedicine offers a promising approach to overcoming this barrier, with nanoparticles (NPs) engineered to deliver therapeutic agents directly to affected brain regions. This review explores the classification and design of NPs, divided into organic and inorganic categories and further categorized based on their chemical and physical properties. These characteristics influence the ability of NPs to carry and release therapeutic agents, target specific tissues and ensure appropriate clearance from the body. The review emphasizes the potential of NPs to enhance the diagnosis and treatment of neurodegenerative diseases through targeted delivery, improved drug bioavailability and real-time therapeutic efficacy monitoring. By addressing the challenges of the compromised BBB and targeting inflammatory biomarkers, NPs represent a cutting-edge strategy in managing neurological disorders, promising better patient outcomes.
Collapse
Affiliation(s)
- Jessica López-Espinosa
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
- School of Medicine and Health Sciences of Tecnológico de Monterrey, Guadalajara, México
| | - Peter Park
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
| | - Morgan Holcomb
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TXUSA
- Department of Obstetrics & Gynecology, Houston Methodist Hospital, Houston, TXUSA
- Department of Obstetrics & Gynecology, Weill Cornell Medicine College, New York, NYUSA
- Department of Biomedical Engineering, Texas A&M University, College Station, TXUSA
| | - Sonia Villapol
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
- Department of Neuroscience in Neurological Surgery, Weill Cornell Medical College, New York, NY USA
| |
Collapse
|
9
|
Zhu L, Zhong W, Meng X, Yang X, Zhang W, Tian Y, Li Y. Polymeric nanocarriers delivery systems in ischemic stroke for targeted therapeutic strategies. J Nanobiotechnology 2024; 22:424. [PMID: 39026255 PMCID: PMC11256638 DOI: 10.1186/s12951-024-02673-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024] Open
Abstract
Ischemic stroke is a complex, high-mortality disease with multifactorial etiology and pathogenesis. Currently, drug therapy is mainly used treat ischemic stroke in clinic, but there are still some limitations, such as limited blood-brain barrier (BBB) penetration efficiency, a narrow treatment time window and drug side effects. Recent studies have pointed out that drug delivery systems based on polymeric nanocarriers can effectively improve the insufficient treatment for ischemic stroke. They can provide neuronal protection by extending the plasma half-life of drugs, enhancing the drug's permeability to penetrate the BBB, and targeting specific structures and cells. In this review, we classified polymeric nanocarriers used for delivering ischemic stroke drugs and introduced their preparation methods. We also evaluated the feasibility and effectiveness and discussed the existing limitations and prospects of polymeric nanocarriers for ischemic stroke treatment. We hoped that this review could provide a theoretical basis for the future development of nanomedicine delivery systems for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Weijie Zhong
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Xuchen Meng
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Xiaosheng Yang
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Wenchuan Zhang
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Yayuan Tian
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China.
| | - Yi Li
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
10
|
Wehn AC, Krestel E, Harapan BN, Klymchenko A, Plesnila N, Khalin I. To see or not to see: In vivo nanocarrier detection methods in the brain and their challenges. J Control Release 2024; 371:216-236. [PMID: 38810705 DOI: 10.1016/j.jconrel.2024.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024]
Abstract
Nanoparticles have a great potential to significantly improve the delivery of therapeutics to the brain and may also be equipped with properties to investigate brain function. The brain, being a highly complex organ shielded by selective barriers, requires its own specialized detection system. However, a significant hurdle to achieve these goals is still the identification of individual nanoparticles within the brain with sufficient cellular, subcellular, and temporal resolution. This review aims to provide a comprehensive summary of the current knowledge on detection systems for tracking nanoparticles across the blood-brain barrier and within the brain. We discuss commonly employed in vivo and ex vivo nanoparticle identification and quantification methods, as well as various imaging modalities able to detect nanoparticles in the brain. Advantages and weaknesses of these modalities as well as the biological factors that must be considered when interpreting results obtained through nanotechnologies are summarized. Finally, we critically evaluate the prevailing limitations of existing technologies and explore potential solutions.
Collapse
Affiliation(s)
- Antonia Clarissa Wehn
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Department of Neurosurgery, University of Munich Medical Center, Marchioninistraße 17, 81377 Munich, Germany.
| | - Eva Krestel
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany.
| | - Biyan Nathanael Harapan
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Department of Neurosurgery, University of Munich Medical Center, Marchioninistraße 17, 81377 Munich, Germany.
| | - Andrey Klymchenko
- Laboratoire de Biophotonique et Pharmacologie, CNRS UMR 7213, Université de Strasbourg, 74 route du Rhin - CS 60024, 67401 Illkirch Cedex, France.
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, 81377 Munich, Germany.
| | - Igor Khalin
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), 14 074 Bd Henri Becquerel, 14000 Caen, France.
| |
Collapse
|
11
|
Susa F, Arpicco S, Pirri CF, Limongi T. An Overview on the Physiopathology of the Blood-Brain Barrier and the Lipid-Based Nanocarriers for Central Nervous System Delivery. Pharmaceutics 2024; 16:849. [PMID: 39065547 PMCID: PMC11279990 DOI: 10.3390/pharmaceutics16070849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The state of well-being and health of our body is regulated by the fine osmotic and biochemical balance established between the cells of the different tissues, organs, and systems. Specific districts of the human body are defined, kept in the correct state of functioning, and, therefore, protected from exogenous or endogenous insults of both mechanical, physical, and biological nature by the presence of different barrier systems. In addition to the placental barrier, which even acts as a linker between two different organisms, the mother and the fetus, all human body barriers, including the blood-brain barrier (BBB), blood-retinal barrier, blood-nerve barrier, blood-lymph barrier, and blood-cerebrospinal fluid barrier, operate to maintain the physiological homeostasis within tissues and organs. From a pharmaceutical point of view, the most challenging is undoubtedly the BBB, since its presence notably complicates the treatment of brain disorders. BBB action can impair the delivery of chemical drugs and biopharmaceuticals into the brain, reducing their therapeutic efficacy and/or increasing their unwanted bioaccumulation in the surrounding healthy tissues. Recent nanotechnological innovation provides advanced biomaterials and ad hoc customized engineering and functionalization methods able to assist in brain-targeted drug delivery. In this context, lipid nanocarriers, including both synthetic (liposomes, solid lipid nanoparticles, nanoemulsions, nanostructured lipid carriers, niosomes, proniosomes, and cubosomes) and cell-derived ones (extracellular vesicles and cell membrane-derived nanocarriers), are considered one of the most successful brain delivery systems due to their reasonable biocompatibility and ability to cross the BBB. This review aims to provide a complete and up-to-date point of view on the efficacy of the most varied lipid carriers, whether FDA-approved, involved in clinical trials, or used in in vitro or in vivo studies, for the treatment of inflammatory, cancerous, or infectious brain diseases.
Collapse
Affiliation(s)
- Francesca Susa
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| | - Candido Fabrizio Pirri
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Tania Limongi
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| |
Collapse
|
12
|
Yuan S, Hu Q. Convergence of nanomedicine and neutrophils for drug delivery. Bioact Mater 2024; 35:150-166. [PMID: 38318228 PMCID: PMC10839777 DOI: 10.1016/j.bioactmat.2024.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024] Open
Abstract
Neutrophils have recently emerged as promising carriers for drug delivery due to their unique properties including rapid response toward inflammation, chemotaxis, and transmigration. When integrated with nanotechnology that has enormous advantages in improving treatment efficacy and reducing side effects, neutrophil-based nano-drug delivery systems have expanded the repertoire of nanoparticles employed in precise therapeutic interventions by either coating nanoparticles with their membranes, loading nanoparticles inside living cells, or engineering chimeric antigen receptor (CAR)-neutrophils. These neutrophil-inspired therapies have shown superior biocompatibility, targeting ability, and therapeutic robustness. In this review, we summarized the benefits of combining neutrophils and nanotechnologies, the design principles and underlying mechanisms, and various applications in disease treatments. The challenges and prospects for neutrophil-based drug delivery systems were also discussed.
Collapse
Affiliation(s)
- Sichen Yuan
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| |
Collapse
|
13
|
Huang T, Wen X, Liang Y, Liu X, Zhao J, Long X. Irreversible Electroporation-Induced Inflammation Facilitates Neutrophil-Mediated Drug Delivery to Enhance Pancreatic Cancer Therapy. Mol Pharm 2024; 21:1998-2011. [PMID: 38412284 DOI: 10.1021/acs.molpharmaceut.4c00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Pancreatic cancer is a deadly disease with a five-year overall survival rate of around 11%. Chemotherapy is a cornerstone in the treatment of this malignancy, but the intratumoral delivery of chemotherapy drugs is impaired by the highly fibrotic tumor-associated stroma. Irreversible electroporation (IRE) is an ablative technique for treating locally advanced pancreatic cancer. During a typical IRE procedure, high-intensity electric pulses are released to kill tumor cells through the irreversible disruption of the cytoplasm membranes. IRE also induces rapid tumor infiltration by neutrophils and offers an opportunity for neutrophil-mediated drug delivery. We herein showed that the IRE-induced neutrophil trafficking was facilitated by the upregulation of neutrophil chemotaxis and migration as well as the release of several chemoattractants. Doxorubicin-loaded bovine serum albumin nanoparticles were prepared and loaded into neutrophils at a ratio of 9.9 ± 1.2 to 11.7 ± 2.0 pg of doxorubicin per cell. The resultant formulation (NP@NEs) efficiently accumulated in the IRE-treated KPC-A377 murine pancreatic tumors with an uptake value of 10.7 ± 1.5 (percent of injected dose per gram of tissue, abbreviated as %ID/g) at 48 h after intravenous injection. In both Panc02 and KPC-A377 murine pancreatic tumor models, the combination of IRE + NP@NEs inhibited tumor growth more effectively than either monotherapy. The tumors treated with the combination also exhibited the lowest frequency of Ki67+ proliferating cells and the highest abundance of terminal deoxynucleotidyl transferase dUTP nick end labeling+ (TUNEL+) apoptotic cells among the experiment groups. Minimal treatment-associated toxicity was observed. Our findings suggest that neutrophil-mediated delivery of chemotherapy drugs is a useful tool to enhance the response of pancreatic cancer to IRE.
Collapse
Affiliation(s)
- Teng Huang
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaofei Wen
- Department of Interventional Radiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 36100, China
- Department of Interventional Radiology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Yuxuan Liang
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiao Liu
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jun Zhao
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xin Long
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
14
|
Zhang SS, Li RQ, Chen Z, Wang XY, Dumont AS, Fan X. Immune cells: potential carriers or agents for drug delivery to the central nervous system. Mil Med Res 2024; 11:19. [PMID: 38549161 PMCID: PMC10979586 DOI: 10.1186/s40779-024-00521-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/05/2024] [Indexed: 04/01/2024] Open
Abstract
Drug delivery systems (DDS) have recently emerged as a promising approach for the unique advantages of drug protection and targeted delivery. However, the access of nanoparticles/drugs to the central nervous system (CNS) remains a challenge mainly due to the obstruction from brain barriers. Immune cells infiltrating the CNS in the pathological state have inspired the development of strategies for CNS foundation drug delivery. Herein, we outline the three major brain barriers in the CNS and the mechanisms by which immune cells migrate across the blood-brain barrier. We subsequently review biomimetic strategies utilizing immune cell-based nanoparticles for the delivery of nanoparticles/drugs to the CNS, as well as recent progress in rationally engineering immune cell-based DDS for CNS diseases. Finally, we discuss the challenges and opportunities of immune cell-based DDS in CNS diseases to promote their clinical development.
Collapse
Affiliation(s)
- Shan-Shan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Ruo-Qi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Zhong Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Xiao-Ying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, 70122, USA
| | - Aaron S Dumont
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, 70122, USA.
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
15
|
Yang J, Shi X, Kuang Y, Wei R, Feng L, Chen J, Wu X. Cell-nanocarrier drug delivery system: a promising strategy for cancer therapy. Drug Deliv Transl Res 2024; 14:581-596. [PMID: 37721694 DOI: 10.1007/s13346-023-01429-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/19/2023]
Abstract
Tumor targeting has been a great challenge for drug delivery systems. A number of nanotechnology-derived drug carriers have been developed for cancer treatment to improve efficacy and biocompatibility. Among them, the emergence of cell-nanocarriers has attracted great attention, which simulates cell function and has good biocompatibility. They can also escape the clearance of reticuloendothelial system, showing a long-cycle effect. The inherent tumor migration and tumor homing ability of cells increase their significance as tumor-targeting vectors. In this review, we focus on the combination of stem cells, immune cells, red blood cells, and cell membranes to nanocarriers, which enable chemotherapy agents to efficiently target lesion sites and improve drug distribution while being low toxic and safe. In addition, we discuss the pros and cons of these nanoparticles as well as the challenges and opportunities that lie ahead. Although research to address these limitations is still ongoing, this promising tumor-targeted drug delivery system will provide a safe and effective platform against cancer.
Collapse
Affiliation(s)
- Jiefen Yang
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China
- Shanghai Wei Er Lab, Shanghai, China
| | - Xiongxi Shi
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China
- Shanghai Wei Er Lab, Shanghai, China
| | - Yanting Kuang
- Shanghai Wei Er Lab, Shanghai, China
- Inner Mongolia Medical University, No. 5, Xinhua Road, Hohhot, Inner Mongolia, People's Republic of China
| | - Ruting Wei
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China
- Shanghai Wei Er Lab, Shanghai, China
| | - Lanni Feng
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China
- Shanghai Wei Er Lab, Shanghai, China
| | - Jianming Chen
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China.
- Shanghai Wei Er Lab, Shanghai, China.
| | - Xin Wu
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China.
- Shanghai Wei Er Lab, Shanghai, China.
| |
Collapse
|
16
|
Lin L, Geng D, She D, Kuai X, Du C, Fu P, Zhu Y, Wang J, Pang Z, Zhang J. Targeted nanotheranostics for the treatment of epilepsy through in vivo hijacking of locally activated macrophages. Acta Biomater 2024; 174:314-330. [PMID: 38036284 DOI: 10.1016/j.actbio.2023.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023]
Abstract
Epilepsy refers to a disabling neurological disorder featured by the long-term and unpredictable occurrence of seizures owing to abnormal excessive neuronal electrical activity and is closely linked to unresolved inflammation, oxidative stress, and hypoxia. The difficulty of accurate localization and targeted drug delivery to the lesion hinders the effective treatment of this disease. The locally activated inflammatory cells in the epileptogenic region offer a new opportunity for drug delivery to the lesion. In this work, CD163-positive macrophages in the epileptogenic region were first harnessed as Trojan horses after being hijacked by targeted albumin manganese dioxide nanoparticles, which effectively penetrated the brain endothelial barrier and delivered multifunctional nanomedicines to the epileptic foci. Hence, accumulative nanoparticles empowered the visualization of the epileptogenic lesion through microenvironment-responsive MR T1-weight imaging of manganese dioxide. Besides, these manganese-based nanomaterials played a pivotal role in shielding neurons from cell apoptosis mediated by oxidative stress and hypoxia. Taken together, the present study provides an up-to-date approach for integrated diagnosis and treatment of epilepsy and other hypoxia-associated inflammatory diseases. STATEMENT OF SIGNIFICANCE: The therapeutic effects of antiepileptic drugs (AEDs) are hindered by insufficient drug accumulation in the epileptic site. Herein, we report an efficient strategy to use locally activated macrophages as carriers to deliver multifunctional nanoparticles to the brain lesion. As MR-responsive T1 contrast agents, multifunctional BMC nanoparticles can be harnessed to accurately localize the epileptogenic region with high sensitivity and specificity. Meanwhile, catalytic nanoparticles BMC can synergistically scavenge ROS, generate O2 and regulate neuroinflammation for the protection of neurons in the brain.
Collapse
Affiliation(s)
- Lin Lin
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China; National Center for Neurological Disorders, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Daoying Geng
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; National Center for Neurological Disorders, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Dejun She
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Xinping Kuai
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Chengjuan Du
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Pengfei Fu
- Department of Neurosurgery, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Yuefei Zhu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery Ministry of Education, Shanghai 201203, China
| | - Jianhong Wang
- National Center for Neurological Disorders, 12 Wulumuqi Middle Road, Shanghai 200040, China; Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China.
| | - Zhiqing Pang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery Ministry of Education, Shanghai 201203, China.
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; National Center for Neurological Disorders, 12 Wulumuqi Middle Road, Shanghai 200040, China.
| |
Collapse
|
17
|
Barmoudeh Z, Fouani MH, Moslemi Z, Azizi M, Doustimotlagh AH, Bardania H. Melatonin and metformin co-loaded nanoliposomes efficiently attenuate liver damage induced by bile duct ligation in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:395-410. [PMID: 37452836 DOI: 10.1007/s00210-023-02613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
In the current study, the therapeutic effectiveness of the metformin (Met) and melatonin (Mel) co-loaded liposomes was investigated on cholestasis induced by bile duct ligation (BDL) in male rats. Histopathological analysis, biochemical analysis, and oxidative stress markers were assayed to determine the therapeutic effect of Met and Mel co-loaded liposomes on cholestasis. Histopathological analysis revealed that the simultaneous administration of Met and Mel, whether in the free (C-Mel-Met) or liposomal (C-Lipo-Mel-Met) forms, reduced inflammation as well as proliferation of bile ducts; however, results were more prominent in the liposomal form of Mel and Met. Additionaly, serum levels of aspartate aminotransferase (AST) were significantly (p < 0.001) higher in (C-Mel-Met) treated rats compared with (BDL) rats; however, (C-Lipo-Mel-Met) treated rats exhibited significant (p < 0.05) lower AST rates in comparison to (BDL) rats. Moreover, a significant (p < 0.0001) drop in bilirubin levels was detected in (C-Lipo-Mel-Met) treated rats in comparison to (BDL) rats; it is noteworthy mentioning that bilirubin levels in (C-Lipo-Mel-Met) treated rats were insignificant in comparison to sham-control (SC) rats. Furthermore, rats concomitantly administered Met and Mel, exhibited significant downregulation in the expression levels of inflammatory cytokine genes such as TNF-α and IL-1 gene expression, where the downregulation was more prominent in the liposomal from. Our findings demonestrate that the concomitant administration of metformin and melatonin in the liposomal form had more therapeutic effect on liver injury than their free forms through improving histological changes, reducing biochemical markers and favoring oxidant- antioxidant balance.
Collapse
Affiliation(s)
- Zahra Barmoudeh
- Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohamad Hassan Fouani
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Moslemi
- Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mahdokht Azizi
- Clinical Research Development Unit, Imamsajad Hospital, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Amir Hossein Doustimotlagh
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| |
Collapse
|
18
|
Hao C, Sha M, Ye Y, Wang C. Cell Membrane-Derived Nanovehicles for Targeted Therapy of Ischemic Stroke: From Construction to Application. Pharmaceutics 2023; 16:6. [PMID: 38276484 PMCID: PMC10819970 DOI: 10.3390/pharmaceutics16010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 01/27/2024] Open
Abstract
Ischemic stroke (IS) is a prevalent form of stroke and a leading cause of mortality and disability. Recently, cell membrane-derived nanovehicles (CMNVs) derived from erythrocytes, thrombocytes, neutrophils, macrophages, neural stem cells, and cancer cells have shown great promise as drug delivery systems for IS treatment. By precisely controlling drug release rates and targeting specific sites in the brain, CMNVs enable the reduction in drug dosage and minimization of side effects, thus significantly enhancing therapeutic strategies and approaches for IS. While there are some reviews regarding the applications of CMNVs in the treatment of IS, there has been limited attention given to important aspects such as carrier construction, structural design, and functional modification. Therefore, this review aims to address these key issues in CMNVs preparation, structural composition, modification, and other relevant aspects, with a specific focus on targeted therapy for IS. Finally, the challenges and prospects in this field are discussed.
Collapse
Affiliation(s)
- Cui Hao
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (H.C.); (S.M.); (Y.Y.)
| | - Ma Sha
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (H.C.); (S.M.); (Y.Y.)
| | - Yang Ye
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (H.C.); (S.M.); (Y.Y.)
- Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan Province, Kunming 650500, China
| | - Chengxiao Wang
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (H.C.); (S.M.); (Y.Y.)
- Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan Province, Kunming 650500, China
| |
Collapse
|
19
|
Liu Z, Xia Q, Ma D, Wang Z, Li L, Han M, Yin X, Ji X, Wang S, Xin T. Biomimetic nanoparticles in ischemic stroke therapy. DISCOVER NANO 2023; 18:40. [PMID: 36969494 PMCID: PMC10027986 DOI: 10.1186/s11671-023-03824-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/07/2023] [Indexed: 05/28/2023]
Abstract
Abstract Ischemic stroke is one of the most severe neurological disorders with limited therapeutic strategies. The utilization of nanoparticle drug delivery systems is a burgeoning field and has been widely investigated. Among these, biomimetic drug delivery systems composed of biogenic membrane components and synthetic nanoparticles have been extensively highlighted in recent years. Biomimetic membrane camouflage presents an effective strategy to prolong circulation, reduce immunogenicity and enhance targeting. For one thing, biomimetic nanoparticles reserve the physical and chemical properties of intrinsic nanoparticle. For another, the biological functions of original source cells are completely inherited. Compared to conventional surface modification methods, this approach is more convenient and biocompatible. In this review, membrane-based nanoparticles derived from different donor cells were exemplified. The prospect of future biomimetic nanoparticles in ischemic stroke therapy was discussed. Graphic abstract
Collapse
Affiliation(s)
- Zihao Liu
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021 China
| | - Qian Xia
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012 China
| | - Dengzhen Ma
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021 China
| | - Zhihai Wang
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021 China
| | - Longji Li
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021 China
| | - Min Han
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 China
| | - Xianyong Yin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 China
| | - Xiaoshuai Ji
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021 China
| | - Shan Wang
- Shandong Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021 Shandong China
| | - Tao Xin
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021 China
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117 China
| |
Collapse
|
20
|
Jiang C, Zhou Y, Chen R, Yang M, Zhou H, Tang Z, Shi H, Qin D. Nanomaterial-Based Drug Delivery Systems for Ischemic Stroke. Pharmaceutics 2023; 15:2669. [PMID: 38140010 PMCID: PMC10748360 DOI: 10.3390/pharmaceutics15122669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Ischemic stroke is a leading cause of death and disability in the world. At present, reperfusion therapy and neuroprotective therapy, as guidelines for identifying effective and adjuvant treatment methods, are limited by treatment time windows, drug bioavailability, and side effects. Nanomaterial-based drug delivery systems have the characteristics of extending half-life, increasing bioavailability, targeting drug delivery, controllable drug release, and low toxicity, thus being used in the treatment of ischemic stroke to increase the therapeutic effects of drugs. Therefore, this review provides a comprehensive overview of nanomaterial-based drug delivery systems from nanocarriers, targeting ligands and stimulus factors of drug release, aiming to find the best combination of nanomaterial-based drug delivery systems for ischemic stroke. Finally, future research areas on nanomaterial-based drug delivery systems in ischemic stroke and the implications of the current knowledge for the development of novel treatment for ischemic stroke were identified.
Collapse
Affiliation(s)
- Chengting Jiang
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China; (C.J.); (M.Y.)
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (R.C.); (H.Z.); (Z.T.)
| | - Yang Zhou
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China;
| | - Rong Chen
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (R.C.); (H.Z.); (Z.T.)
| | - Mengjia Yang
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China; (C.J.); (M.Y.)
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (R.C.); (H.Z.); (Z.T.)
| | - Haimei Zhou
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (R.C.); (H.Z.); (Z.T.)
| | - Zhengxiu Tang
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (R.C.); (H.Z.); (Z.T.)
| | - Hongling Shi
- Department of Rehabilitation Medicine, The Affiliated Hospital of Yunnan University, Kunming 650021, China
| | - Dongdong Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China; (C.J.); (M.Y.)
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (R.C.); (H.Z.); (Z.T.)
| |
Collapse
|
21
|
Yu X, Luo Y, Yang L, Duan X. Plasma metabonomic study on the effect of Para‑hydroxybenzaldehyde intervention in a rat model of transient focal cerebral ischemia. Mol Med Rep 2023; 28:224. [PMID: 37800608 PMCID: PMC10577806 DOI: 10.3892/mmr.2023.13111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/28/2023] [Indexed: 10/07/2023] Open
Abstract
Gastrodia elata Blume has been widely used to treat various central and peripheral nerve diseases, and Para‑hydroxybenzaldehyde (PHBA) is one of the indicated components suggested to provide a neuroprotective effect. In our previous, it was shown that PHBA protected mitochondria against cerebral ischemia‑reperfusion (I/R) injury in rats. In the present study, how PHBA regulated the metabolic mechanism in blood following cerebral I/R was assessed to identify an effective therapeutic target for the prevention and treatment of ischemic stroke (IS). First, a rat model of cerebral ischemia‑reperfusion injury was established via middle cerebral artery occlusion/reperfusion (MCAO/R). The therapeutic effect of PHBA on brain I/R was evaluated by assessing the neurological function score, triphenyl tetrazolium chloride, hematoxylin and eosin, and Nissl staining. Next, a non‑targeted metabolomic based on high‑performance liquid chromatography quadrupole time‑of‑flight mass spectrometry was established to identify differential metabolites. Finally, a targeted metabolic spectrum was analyzed and the potential therapeutic targets were verified by Western blotting. The results showed that the neurological function score, cerebral infarction area, hippocampal morphology, and the number of neurons in the PHBA group were significantly improved compared with the model group. Metabonomic analysis showed that 13 different metabolites were identified between the model and PHBA group, which may be involved in the 'tricarboxylic acid cycle', 'glutathione metabolism', and 'mutual transformation of pentose and glucuronates', amongst others. Among these, the levels of the most significant differential metabolite, dGMP, decreased significantly following PHBA treatment. Western blotting was used to verify the expression of membrane‑associated guanosine kinase PSD‑95 and the subunit of glutamate AMPA receptor GluA1, which significantly increased after PHBA treatment. In addition, it was also found that PHBA increased the expression of the light chain‑3 protein and autophagy effector protein 1, whilst the expression of sequestosome‑1 decreased, indicating that PHBA promoted autophagy. Similarly, in TUNEL staining and detection of apoptosis‑related proteins, it was found that MCAO/R upregulated the expression of Bax and cleaved‑caspase‑3 whilst downregulating the expression of Bcl‑2 and increasing the apoptosis of hippocampal neurons; PHBA reversed this situation. These results suggest that cerebral I/R causes postsynaptic dysfunction by disrupting the interaction between PSD‑95 and AMPARs, and the inhibition of the autophagy system eventually leads to the apoptosis of hippocampal neurons.
Collapse
Affiliation(s)
- Xinglin Yu
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yuan Luo
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Liping Yang
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Xiaohua Duan
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
22
|
Nong J, Glassman PM, Myerson JW, Zuluaga-Ramirez V, Rodriguez-Garcia A, Mukalel A, Omo-Lamai S, Walsh LR, Zamora ME, Gong X, Wang Z, Bhamidipati K, Kiseleva RY, Villa CH, Greineder CF, Kasner SE, Weissman D, Mitchell MJ, Muro S, Persidsky Y, Brenner JS, Muzykantov VR, Marcos-Contreras OA. Targeted Nanocarriers Co-Opting Pulmonary Intravascular Leukocytes for Drug Delivery to the Injured Brain. ACS NANO 2023; 17:13121-13136. [PMID: 37432926 PMCID: PMC10373654 DOI: 10.1021/acsnano.2c08275] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 06/08/2023] [Indexed: 07/13/2023]
Abstract
Ex vivo-loaded white blood cells (WBC) can transfer cargo to pathological foci in the central nervous system (CNS). Here we tested affinity ligand driven in vivo loading of WBC in order to bypass the need for ex vivo WBC manipulation. We used a mouse model of acute brain inflammation caused by local injection of tumor necrosis factor alpha (TNF-α). We intravenously injected nanoparticles targeted to intercellular adhesion molecule 1 (anti-ICAM/NP). We found that (A) at 2 h, >20% of anti-ICAM/NP were localized to the lungs; (B) of the anti-ICAM/NP in the lungs >90% were associated with leukocytes; (C) at 6 and 22 h, anti-ICAM/NP pulmonary uptake decreased; (D) anti-ICAM/NP uptake in brain increased up to 5-fold in this time interval, concomitantly with migration of WBCs into the injured brain. Intravital microscopy confirmed transport of anti-ICAM/NP beyond the blood-brain barrier and flow cytometry demonstrated complete association of NP with WBC in the brain (98%). Dexamethasone-loaded anti-ICAM/liposomes abrogated brain edema in this model and promoted anti-inflammatory M2 polarization of macrophages in the brain. In vivo targeted loading of WBC in the intravascular pool may provide advantages of coopting WBC predisposed to natural rapid mobilization from the lungs to the brain, connected directly via conduit vessels.
Collapse
Affiliation(s)
- Jia Nong
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Patrick M. Glassman
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department
of Pharmaceutical Sciences, Temple University
School of Pharmacy, Philadelphia, Pennsylvania 19140, United States
| | - Jacob W. Myerson
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Viviana Zuluaga-Ramirez
- Department
of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Alba Rodriguez-Garcia
- Department
of Pathology and Laboratory Medicine, Ovarian Cancer Research Center,
Perelman School of Medicine, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center
for Cellular Immunotherapies, Abramson Cancer Center, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Alvin Mukalel
- Department
of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Serena Omo-Lamai
- Division
of Pulmonary Allergy, and Critical Care, Department of Medicine, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Landis R. Walsh
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Marco E. Zamora
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- School
of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Xijing Gong
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Division
of Pulmonary Allergy, and Critical Care, Department of Medicine, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Zhicheng Wang
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Kartik Bhamidipati
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Raisa Y. Kiseleva
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Carlos H. Villa
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Colin Fred Greineder
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Scott E. Kasner
- Department
of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Drew Weissman
- Division
of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael J. Mitchell
- Department
of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Abramson
Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute
for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular
Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute
for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, 08028, Spain
- Institute of Catalonia for Research and
Advanced Studies (ICREA), Barcelona, 08010, Spain
- Institute
for Bioscience and Biotechnology (IBBR), College Park, Maryland 20850, United States
| | - Yuri Persidsky
- Department
of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
- Center
for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Jacob Samuel Brenner
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Division
of Pulmonary Allergy, and Critical Care, Department of Medicine, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R. Muzykantov
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A. Marcos-Contreras
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department
of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
23
|
Zhao Y, Zhang H, Zhang Q, Tao H. Research Progress of Neutrophil-Mediated Drug Delivery Strategies for Inflammation-Related Disease. Pharmaceutics 2023; 15:1881. [PMID: 37514067 PMCID: PMC10384340 DOI: 10.3390/pharmaceutics15071881] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
As the most abundant white blood cells in humans, neutrophils play a key role in acute and chronic inflammation, suggesting that these cells are a key component of targeted therapies for various inflammation-related diseases. Specific enzyme-responsive or specific ligand-modified polymer nanoparticles are beneficial for improving drug efficacy, reducing toxicity, and enhancing focal site retention. However, there remain significant challenges in biomedical applications of these synthetic polymer nanoparticles, mainly due to their rapid clearance by the reticuloendothelial system. In recent years, biomimetic drug delivery systems such as neutrophils acting directly as drug carriers or neutrophil-membrane-coated nanoparticles have received increasing attention due to the natural advantages of neutrophils. Thus, neutrophil-targeted, neutrophil-assisted, or neutrophil-coated nanoparticles exhibit a prolonged blood circulation time and improved accumulation at the site of inflammation. Despite recent advancements, further clinical research must be performed to evaluate neutrophil-based delivery systems for future biomedical application in the diagnosis and treatment of related inflammatory diseases. In this review, we have summarized new exciting developments and challenges in neutrophil-mediated drug delivery strategies for treating inflammation-related diseases.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Pharmaceutics, 96602 Hospital of Chinese People's Liberation Army, Kunming 650233, China
| | - Haigang Zhang
- Department of Pharmacology, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Qixiong Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Innovation Center of Advanced Pharmaceutical & Artificial Intelligence, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Hui Tao
- Department of Pharmacology, College of Pharmacy, Army Medical University, Chongqing 400038, China
| |
Collapse
|
24
|
Mu Q, Yao K, Syeda MZ, Zhang M, Cheng Q, Zhang Y, Sun R, Lu Y, Zhang H, Luo Z, Huang H, Liu X, Luo C, Zhu X, Wu S, Cui L, Huang C, Chen X, Tang L. Ligustrazine Nanoparticle Hitchhiking on Neutrophils for Enhanced Therapy of Cerebral Ischemia-Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301348. [PMID: 37078794 PMCID: PMC10323616 DOI: 10.1002/advs.202301348] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/19/2023] [Indexed: 05/03/2023]
Abstract
Ischemic stroke is a refractory disease that endangers human health and safety owing to cerebral ischemia. Brain ischemia induces a series of inflammatory reactions. Neutrophils migrate from the circulatory system to the site of cerebral ischemia and accumulate in large numbers at the site of inflammation across the blood-brain barrier. Therefore, hitchhiking on neutrophils to deliver drugs to ischemic brain sites could be an optimal strategy. Since the surface of neutrophils has a formyl peptide receptor (FPR), this work modifies a nanoplatform surface by the peptide cinnamyl-F-(D)L-F-(D)L-F (CFLFLF), which can specifically bind to the FPR receptor. After intravenous injection, the fabricated nanoparticles effectively adhered to the surface of neutrophils in peripheral blood mediated by FPR, thereby hitchhiking with neutrophils to achieve higher accumulation at the inflammatory site of cerebral ischemia. In addition, the nanoparticle shell is composed of a polymer with reactive oxygen species (ROS)-responsive bond breaking and is encased in ligustrazine, a natural product with neuroprotective properties. In conclusion, the strategy of hitching the delivered drugs to neutrophils in this study could improve drug enrichment in the brain, thereby providing a general delivery platform for ischemic stroke or other inflammation-related diseases.
Collapse
Affiliation(s)
- Qingchun Mu
- The People's Hospital of GaozhouGuangdong Medical UniversityMaoming525200China
| | - Kai Yao
- Department of NeurosurgeryFirst Affiliated Hospital of Harbin Medical UniversityHarbin150001China
| | - Madiha Zahra Syeda
- The People's Hospital of GaozhouGuangdong Medical UniversityMaoming525200China
| | - Min Zhang
- International Institutes of MedicineThe Fourth Affiliated HospitalZhejiang University School of MedicineYiwu322000China
| | - Qian Cheng
- Basic Medical CollegeGuilin Medical UniversityGuilin541199China
| | - Yufei Zhang
- Basic Medical CollegeGuilin Medical UniversityGuilin541199China
| | - Rui Sun
- School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Yuting Lu
- International Institutes of MedicineThe Fourth Affiliated HospitalZhejiang University School of MedicineYiwu322000China
| | - Huamiao Zhang
- International Institutes of MedicineThe Fourth Affiliated HospitalZhejiang University School of MedicineYiwu322000China
| | - Zhicheng Luo
- The People's Hospital of GaozhouGuangdong Medical UniversityMaoming525200China
| | - Hanning Huang
- The People's Hospital of GaozhouGuangdong Medical UniversityMaoming525200China
| | - Xiaojing Liu
- The People's Hospital of GaozhouGuangdong Medical UniversityMaoming525200China
| | - Chunmei Luo
- The People's Hospital of GaozhouGuangdong Medical UniversityMaoming525200China
| | - Xiulong Zhu
- The People's Hospital of GaozhouGuangdong Medical UniversityMaoming525200China
| | - Shuyu Wu
- Department of NeurosurgeryHainan General HospicalHainan Affiliated Hospital of Hainan Medical UniversityHaikou570311China
| | - Liao Cui
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs and School of PharmacyGuangdong Medical UniversityDongguan523808China
| | - Chunming Huang
- The People's Hospital of GaozhouGuangdong Medical UniversityMaoming525200China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiologyand SurgeryClinical Imaging Research CentreCentre for Translational MedicineNanomedicine Translational Research ProgramNUS Center for NanomedicineYong Loo Lin School of MedicineDepartments of Chemical and Biomolecular Engineeringand Biomedical EngineeringFaculty of EngineeringNational University of SingaporeSingapore117597Singapore
| | - Longguang Tang
- The People's Hospital of GaozhouGuangdong Medical UniversityMaoming525200China
| |
Collapse
|
25
|
Wang J, Wang Y, Xiaohalati X, Su Q, Liu J, Cai B, Yang W, Wang Z, Wang L. A Bioinspired Manganese-Organic Framework Ameliorates Ischemic Stroke through its Intrinsic Nanozyme Activity and Upregulating Endogenous Antioxidant Enzymes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206854. [PMID: 37129343 PMCID: PMC10369237 DOI: 10.1002/advs.202206854] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/21/2023] [Indexed: 05/03/2023]
Abstract
Following stroke, oxidative stress induced by reactive oxygen species (ROS) aggravates neuronal damage and enlarges ischemic penumbra, which is devastating to stroke patients. Nanozyme-based antioxidants are emerging to treat stroke through scavenging excessive ROS. However, most of nanozymes cannot efficiently scavenge ROS in neuronal cytosol and mitochondria, due to low-uptake abilities of neurons and barriers of organelle membranes, significantly limiting nanozymes' neuroprotective effects. To overcome this limitation, a manganese-organic framework modified with polydopamine (pDA-MNOF), capable of not only mimicking catalytic activities of natural SOD2's catalytic domain but also upregulating two endogenous antioxidant enzymes in neurons is fabricated. With such a dual anti-ROS effect, this nanozyme robustly decreases cellular ROS and effectively protects them from ROS-induced injury. STAT-3 signaling is found to play a vital role in pDA-MNOF activating the two antioxidant enzymes, HO1 and SOD2. In vivo pDA-MNOF treatment significantly improves the survival of middle cerebral artery occlusion (MCAo) mice by reducing infarct volume and more importantly, promotes animal behavioral recovery. Further, pDA-MNOF activates vascular endothelial growth factor expression, a downstream target of STAT3 signaling, thus enhancing angiogenesis. Taken together, the biochemical, cell-biological, and animal-level behavioral data demonstrate the potentiality of pDA-MNOF as a dual ROS-scavenging agent for stroke treatment.
Collapse
Affiliation(s)
- Jian Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Yang Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Xiakeerzhati Xiaohalati
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Qiangfei Su
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Jingwei Liu
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Bo Cai
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Wen Yang
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Zheng Wang
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Lin Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| |
Collapse
|
26
|
Wang K, Zhou W, Jin X, Shang X, Wu X, Wen L, Li S, Hong Y, Ke J, Xu Y, Yuan H, Hu F. Enhanced brain delivery of hypoxia-sensitive liposomes by hydroxyurea for rescue therapy of hyperacute ischemic stroke. NANOSCALE 2023. [PMID: 37377137 DOI: 10.1039/d3nr01071f] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Ischemic stroke is characterized by high morbidity, disability, and mortality. Unfortunately, the only FDA-approved pharmacological thrombolytic, alteplase, has a narrow therapeutic window of only 4.5 h. Other drugs like neuroprotective agents have not been clinically used because of their low efficacy. To improve the efficacy of neuroprotective agents and the effectiveness of rescue therapies for hyperacute ischemic stroke, we investigated and verified the variation trends of the blood-brain barrier (BBB) permeability and regional cerebral blood flow over 24 h in rats that had ischemic strokes. Hypoperfusion and the biphasic increase of BBB permeability are still the main limiting factors for lesion-specific drug distribution and drug brain penetration. Herein, the nitric oxide donor hydroxyurea (HYD) was reported to downregulate the expression of tight junction proteins and upregulate intracellular nitric oxide content in the brain microvascular endothelial cells subjected to oxygen-glucose deprivation, which was shown to facilitate the transport of liposomes across brain endothelial monolayer in an in vitro model. HYD also increased the BBB permeability and promoted microcirculation in the hyperacute phase of stroke. The neutrophil-like cell-membrane-fusogenic hypoxia-sensitive liposomes exhibited excellent performance in targeting the inflamed brain microvascular endothelial cells, enhancing cell association, and promoting rapid hypoxic-responsive release in the hypoxic microenvironment. Overall, the combined HYD and hypoxia-sensitive liposome dosing regimen effectively decreased the cerebral infarction volume and relieved neurological dysfunction in rats that had ischemic strokes; these therapies were involved in the anti-oxidative stress effect and the neurotrophic effect mediated by macrophage migration inhibitory factor.
Collapse
Affiliation(s)
- Kai Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Wentao Zhou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Xiangyu Jin
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xuwei Shang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Xiaomei Wu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Lijuan Wen
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Sufen Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Yiling Hong
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Jia Ke
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Yichong Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Fuqiang Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| |
Collapse
|
27
|
Zhong X, Sun Y, Lu Y, Xu L. Immunomodulatory role of estrogen in ischemic stroke: neuroinflammation and effect of sex. Front Immunol 2023; 14:1164258. [PMID: 37180115 PMCID: PMC10167039 DOI: 10.3389/fimmu.2023.1164258] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023] Open
Abstract
Although estrogen is predominantly related to the maintenance of reproductive functioning in females, it mediates various physiological effects in nearly all tissues, especially the central nervous system. Clinical trials have revealed that estrogen, especially 17β-estradiol, can attenuate cerebral damage caused by an ischemic stroke. One mechanism underlying this effect of 17β-estradiol is by modulating the responses of immune cells, indicating its utility as a novel therapeutic strategy for ischemic stroke. The present review summarizes the effect of sex on ischemic stroke progression, the role of estrogen as an immunomodulator in immune reactions, and the potential clinical value of estrogen replacement therapy. The data presented here will help better understand the immunomodulatory function of estrogen and may provide a basis for its novel therapeutic use in ischemic stroke.
Collapse
Affiliation(s)
- Xiaojun Zhong
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yulin Sun
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yajun Lu
- Department of Internal Medicine, Sunto Women & Children’s Hospital, Jiaxing, China
| | - Lei Xu
- Department of Neurology, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|
28
|
Ou A, Wang Y, Zhang J, Huang Y. Living Cells and Cell-Derived Vesicles: A Trojan Horse Technique for Brain Delivery. Pharmaceutics 2023; 15:pharmaceutics15041257. [PMID: 37111742 PMCID: PMC10145830 DOI: 10.3390/pharmaceutics15041257] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Brain diseases remain a significant global healthcare burden. Conventional pharmacological therapy for brain diseases encounters huge challenges because of the blood-brain barrier (BBB) limiting the delivery of therapeutics into the brain parenchyma. To address this issue, researchers have explored various types of drug delivery systems. Cells and cell derivatives have attracted increasing interest as "Trojan horse" delivery systems for brain diseases, owing to their superior biocompatibility, low immunogenicity, and BBB penetration properties. This review provided an overview of recent advancements in cell- and cell-derivative-based delivery systems for the diagnosis and treatment of brain diseases. Additionally, it discussed the challenges and potential solutions for clinical translation.
Collapse
Affiliation(s)
- Ante Ou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuewei Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaxin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
29
|
Zhou L, Wu X, Qin S, Shi J, Yu C, Xu Z, Tian G, Zhu W, Qin J. Cell-liposome delivery system based on neuroinflammation to target the amygdala for ameliorating depressive-like behaviors. Int J Pharm 2023; 637:122724. [PMID: 36958607 DOI: 10.1016/j.ijpharm.2023.122724] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 03/25/2023]
Abstract
Depression is a serious psychiatric disorder with unsatisfactory outcomes due to difficulties in delivering therapeutic molecules from the periphery to the brain. Neuroinflammation plays a key role in neurobiology and the treatment of depression. Neutrophils can cross the blood-brain barrier (BBB) and infiltrate key brain regions related to the pathophysiology of depression during neuroinflammation. N-Acetyl Pro-Gly-Pro (PGP) peptides efficiently bind to CXCR2 receptors on the surface of neutrophils. The neuropeptide oxytocin demonstrated antidepressant properties in preclinical and clinical studies, but its inability to penetrate the BBB hampers its therapeutic applications. In this study, we established a novel drug delivery system based on neutrophil infiltration in key brain regions during neuroinflammation. PGP was used to modify oxytocin-loaded liposomes (PGP-OTL) as the target ligand. Systematic administration of PGP-OTL exhibited enhanced antidepressant properties resulting from elevated oxytocin concentrations, especially in the amygdala, a crucial depression-implicated brain region. Enhanced antidepressant effects of PGP-OTL, similar to the ones caused by central oxytocin infusion, were observed in behavioral measurement including forced swim and tail suspension tests. Our study demonstrated that PGP-OTL can "hitchhike" neutrophils and enhance delivery of therapeutics into the brain, thus providing the means for developing novel cell-liposome-based drug delivery strategies for depression therapy.
Collapse
Affiliation(s)
- Liping Zhou
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Xiao Wu
- National Institute on Drug Dependence & Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Sijie Qin
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jing Shi
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Chunfeng Yu
- Department of Pharmaceutics, School of Pharmacy, Heilongjiang University of Chinese Medicine
| | - Zhaowei Xu
- Precision Medicine Research Center, School of Pharmacy, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Geng Tian
- Precision Medicine Research Center, School of Pharmacy, Binzhou Medical University, Yantai, Shandong Province 264003, China.
| | - Weili Zhu
- National Institute on Drug Dependence & Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China.
| | - Jing Qin
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China.
| |
Collapse
|
30
|
Sun X, Li M, Huang S, Zhang H, Li K. Protective effect of bone morphogenetic protein-7 induced differentiation of bone marrow mesenchymal stem cells in rat with acute spinal cord injury. Funct Integr Genomics 2023; 23:68. [PMID: 36849554 DOI: 10.1007/s10142-023-00994-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/08/2023] [Accepted: 02/17/2023] [Indexed: 03/01/2023]
Abstract
The principal aim of present study was to assess the therapeutic efficacy of bone morphogenetic protein-7 (BMP-7) induced differentiation of bone marrow mesenchymal stem cells (BMSCs) in a rat acute spinal cord injury (SCI) model. BMSCs were isolated from rats, and then divided into a control and a BMP-7 induction groups. The proliferation ability of BMSCs and glial cell markers were determined. Forty Sprague-Dawley (SD) rats were randomly divided into sham, SCI, BMSC, and BMP7 + BMSC groups (n = 10). Among these rats, the recovery of hind limb motor function, the pathological related markers, and motor evoked potentials (MEP) were identified. BMSCs differentiated into neuron-like cells after the introduction of exogenous BMP-7. Interestingly, the expression levels of MAP-2 and Nestin increased, whereas the expression level of GFAP decreased after the treatment with exogenous BMP-7. Furthermore, the Basso, Beattie, and Bresnahan (BBB) score reached 19.33 ± 0.58 in the BMP-7 + BMSC group at day 42. Nissl bodies in the model group were reduced compared to the sham group. After 42 days, in both the BMSC and BMP-7 + BMSC groups, the number of Nissl bodies increased. This is especially so for the number of Nissl bodies in the BMP-7 + BMSC group, which was more than that in the BMSC group. The expression of Tuj-1 and MBP in BMP-7 + BMSC group increased, whereas the expression of GFAP decreased. Moreover, the MEP waveform decreased significantly after surgery. Furthermore, the waveform was wider and the amplitude was higher in BMP-7 + BMSC group than that in BMSC group. BMP-7 promotes BMSC proliferation, induces the differentiation of BMSCsinto neuron-like cells, and inhibits the formation of glial scar. BMP-7 plays a confident role in the recovery of SCI rats.
Collapse
Affiliation(s)
- Xudong Sun
- The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.280, Changhuai Road, Longzihu Distract, Bengbu, 233044, Anhui Province, China
| | - Maoyong Li
- The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.280, Changhuai Road, Longzihu Distract, Bengbu, 233044, Anhui Province, China
| | - Shiyuan Huang
- The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.280, Changhuai Road, Longzihu Distract, Bengbu, 233044, Anhui Province, China
| | - Heng Zhang
- The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.280, Changhuai Road, Longzihu Distract, Bengbu, 233044, Anhui Province, China.
| | - Kuanxin Li
- The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.280, Changhuai Road, Longzihu Distract, Bengbu, 233044, Anhui Province, China.
| |
Collapse
|
31
|
Cell-Membrane-Coated Nanoparticles for Targeted Drug Delivery to the Brain for the Treatment of Neurological Diseases. Pharmaceutics 2023; 15:pharmaceutics15020621. [PMID: 36839943 PMCID: PMC9960717 DOI: 10.3390/pharmaceutics15020621] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/21/2023] [Accepted: 02/01/2023] [Indexed: 02/16/2023] Open
Abstract
Neurological diseases (NDs) are a significant cause of disability and death in the global population. However, effective treatments still need to be improved for most NDs. In recent years, cell-membrane-coated nanoparticles (CMCNPs) as drug-targeting delivery systems have become a research hotspot. Such a membrane-derived, nano drug-delivery system not only contributes to avoiding immune clearance but also endows nanoparticles (NPs) with various cellular and functional mimicries. This review article first provides an overview of the function and mechanism of single/hybrid cell-membrane-derived NPs. Then, we highlight the application and safety of CMCNPs in NDs. Finally, we discuss the challenges and opportunities in the field.
Collapse
|
32
|
Cardiovascular Nanotechnology. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
33
|
Jiang Y, Liu Z, Liao Y, Sun S, Dai Y, Tang Y. Ischemic stroke: From pathological mechanisms to neuroprotective strategies. Front Neurol 2022; 13:1013083. [PMID: 36438975 PMCID: PMC9681807 DOI: 10.3389/fneur.2022.1013083] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
Ischemic stroke (IS) has complex pathological mechanisms, and is extremely difficult to treat. At present, the treatment of IS is mainly based on intravenous thrombolysis and mechanical thrombectomy, but they are limited by a strict time window. In addition, after intravenous thrombolysis or mechanical thrombectomy, damaged neurons often fail to make ideal improvements due to microcirculation disorders. Therefore, finding suitable pathways and targets from the pathological mechanism is crucial for the development of neuroprotective agents against IS. With the hope of making contributions to the development of IS treatments, this review will introduce (1) how related targets are found in pathological mechanisms such as inflammation, excitotoxicity, oxidative stress, and complement system activation; and (2) the current status and challenges in drug development.
Collapse
Affiliation(s)
- Yang Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenquan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Liao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuyong Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yajie Dai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yibo Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
34
|
Liu T, Gao C, Gu D, Tang H. Cell-based carrier for targeted hitchhiking delivery. Drug Deliv Transl Res 2022; 12:2634-2648. [PMID: 35499717 DOI: 10.1007/s13346-022-01149-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 12/15/2022]
Abstract
Drug delivery systems aim at improving drug transport efficiency and therapeutic efficacy by rational design, and current research on conventional delivery systems brings new developments for disease treatment. Recently, studies on cell-based drug delivery systems are rapidly emerging, which shows great advantages in comparison to conventional drug delivery system. The system uses cells as carriers to delivery conventional drugs or nanomedicines and shows good biocompatibility and enhanced targeting efficiency, beneficial from self component and its physiological function. The construction methodology of cell-based carrier determines the effect on the physiological functions of transporting cell and affects its clinical application. There are different strategies to prepare cell-based carrier, such as direct internalization or surface conjugation of drugs or drug loaded materials. Thus, it is necessary to fully understand the advantages and disadvantages of different strategies for constructing cell-based carrier and then to seek the appropriate construction methodology for achieving better therapeutic results based on disease characterization. We here summarize the application of different types of cell-based carriers reported in recent years and further discuss their applications in disease therapy and the dilemmas faced in clinical translation. We hope that this summary can accelerate the process of clinical translation by promoting the technology development of cell-based carrier.
Collapse
Affiliation(s)
- Tonggong Liu
- Department of Preventive Medicine, School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, China.,Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Cheng Gao
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China.,Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dayong Gu
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Huanwen Tang
- Department of Preventive Medicine, School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
35
|
Wei C, Shen T, Tang X, Gao Y, Yu X, Chen X. Cerebral small vessel disease combined with cerebral collaterals to predict the prognosis of patients with acute large artery atherosclerotic stroke. Front Neurol 2022; 13:969637. [PMID: 36034278 PMCID: PMC9403757 DOI: 10.3389/fneur.2022.969637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Background and purpose Besides cerebral collaterals, few studies have examined other additional factors affecting the prognosis of patients with large artery atherosclerotic (LAA) stroke. Our study aims to explore the effect of the cerebral small vessel disease (SVD) and the effects of its interaction with cerebral collaterals on the prognosis of patients with acute LAA stroke. Method Patients aged 18 years or older with LAA stroke within 24 h after stroke onset were consecutively enrolled. The functional outcome was determined using the modified Rankin Scale (mRS) at 3 months after stroke onset. Logistic multivariate analyses were used to identify the risk factors for stroke prognosis. Receiver operating characteristic (ROC) curves were constructed to compare the effects of cerebral collaterals and SVD on predicting the prognosis. Results Of the 274 enrolled patients, 174 (63.50%) were identified as having a favorable prognosis, and 100 (36.50%) were identified as having an unfavorable prognosis. After adjusting for covariates, the logistic regression analysis identified that unfavorable prognosis was related to the total SVD score (Model 1, adjusted odds ratio = 1.73, 95% CI: 1.15–2.61, P < 0.01; Model 2, adjusted odds ratio = 1.85, 95% CI: 1.23–2.79, P < 0.01) and Tan score (Model 1, adjusted odds ratio = 0.38, 95% CI: 0.23–0.64, P < 0.01; Model 2, adjusted odds ratio = 0.52, 95% CI: 0.33–0.82, P < 0.01). Compared with cerebral collaterals (AUC = 0.59; 95% CI: 0.52–0.67; P < 0.01) or SVD (AUC = 0.62; 95% CI: 0.56–0.69; P < 0.01) alone, the combination of collaterals and SVD (AUC = 0.66; 95% CI: 0.59–0.73; P < 0.01) had higher diagnostic value for an unfavorable prognosis, and the optimal sensitivity and specificity were 77.01 and 53.00%, respectively. Conclusions The total SVD burden was related to the prognosis of patients with LAA stroke. Compared with cerebral collaterals or SVD alone, cerebral collaterals combined with total SVD burden are better at predicting the prognosis of patients with acute LAA stroke.
Collapse
Affiliation(s)
- Cunsheng Wei
- Department of Neurology, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
| | - Tingwen Shen
- The Health Promotion Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuelian Tang
- Department of Neurology, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
| | - Yuanyuan Gao
- Department of General Practice, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
| | - Xiaorong Yu
- Department of Neurology, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
| | - Xuemei Chen
- Department of Neurology, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
- *Correspondence: Xuemei Chen
| |
Collapse
|
36
|
Wang Q, Chen Y, Meng L, Yin J, Wang L, Gong T. A Novel Perspective on Ischemic Stroke: A Review of Exosome and Noncoding RNA Studies. Brain Sci 2022; 12:1000. [PMID: 36009062 PMCID: PMC9406049 DOI: 10.3390/brainsci12081000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/27/2022] [Accepted: 07/18/2022] [Indexed: 02/01/2023] Open
Abstract
Ischemic stroke is a life-threatening condition that also frequently results in long-term disability. Currently, intravenous thrombolysis with tissue plasminogen activator and mechanical thrombectomy is the most popular treatment. However, the narrow time window and related complications limit the treatment benefits. Exosomes have recently emerged as ideal therapeutic candidates for ischemic stroke with the ability to pass through the blood_brain barrier and mediate intercellular communication, in addition, exosomes and their contents can be bioengineered to implement targeted delivery. In the last two decades, exosomes and exosomal noncoding RNAs have been found to be involved in the pathophysiological progression of ischemic stroke, including atherosclerosis, apoptosis, inflammation, oxidative stress, and neurovascular remodeling. In this review, we describe the latest progress regarding the role of exosomal long noncoding RNAs and circular RNAs in the occurrence, progression, and recovery of ischemic stroke. Exploration of exosomal noncoding RNAs and their correlated effects in ischemic stroke may facilitate accurate diagnosis, and they may serve as new therapeutic targets for the disease.
Collapse
Affiliation(s)
- Qianwen Wang
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing 100730, China; (Q.W.); (Y.C.); (J.Y.); (L.W.)
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdansantiao, Dongcheng District, Beijing 100730, China;
| | - Yuhui Chen
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing 100730, China; (Q.W.); (Y.C.); (J.Y.); (L.W.)
| | - Lingbing Meng
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdansantiao, Dongcheng District, Beijing 100730, China;
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing 100730, China
| | - Jiawen Yin
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing 100730, China; (Q.W.); (Y.C.); (J.Y.); (L.W.)
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdansantiao, Dongcheng District, Beijing 100730, China;
| | - Li Wang
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing 100730, China; (Q.W.); (Y.C.); (J.Y.); (L.W.)
| | - Tao Gong
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing 100730, China; (Q.W.); (Y.C.); (J.Y.); (L.W.)
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdansantiao, Dongcheng District, Beijing 100730, China;
| |
Collapse
|
37
|
Lv W, Liu Y, Li S, Lv L, Lu H, Xin H. Advances of nano drug delivery system for the theranostics of ischemic stroke. J Nanobiotechnology 2022; 20:248. [PMID: 35641956 PMCID: PMC9153106 DOI: 10.1186/s12951-022-01450-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 05/05/2022] [Indexed: 02/07/2023] Open
Abstract
From the global perspective, stroke refers to a highly common cause of disability and death. Ischemic stroke (IS), attributed to blood vessel blockage, preventing the flow of blood to brain, acts as the most common form of stroke. Thus far, thrombolytic therapy is the only clinical treatment for IS with the approval from the FDA. Moreover, the physiology barrier complicates therapeutically and diagnostically related intervention development of IS. Accordingly, developing efficient and powerful curative approaches for IS diagnosis and treatment is urgently required. The advent of nanotechnology has brought dawn and hope to better curative and imaging forms for the management of IS. This work reviews the recent advances and challenges correlated with the nano drug delivery system for IS therapy and diagnosis. The overview of the current knowledge of the important molecular pathological mechanisms in cerebral ischemia and how the drugs cross the blood brain barrier will also be briefly summarized.
Collapse
Affiliation(s)
- Wei Lv
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, 214400, Jiangyin, China
| | - Yijiao Liu
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, 214400, Jiangyin, China
| | - Shengnan Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China
| | - Lingyan Lv
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, 214400, Jiangyin, China
| | - Hongdan Lu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China.
| | - Hongliang Xin
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China.
| |
Collapse
|
38
|
An J, Zhao L, Duan R, Sun K, Lu W, Yang J, Liang Y, Liu J, Zhang Z, Li L, Shi J. Potential nanotherapeutic strategies for perioperative stroke. CNS Neurosci Ther 2022; 28:510-520. [PMID: 35243774 PMCID: PMC8928924 DOI: 10.1111/cns.13819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 12/12/2022] Open
Abstract
AIMS Based on the complex pathological environment of perioperative stroke, the development of targeted therapeutic strategies is important to control the development of perioperative stroke. DISCUSSIONS Recently, great progress has been made in nanotechnology, and nanodrug delivery systems have been developed for the treatment of ischemic stroke. CONCLUSION In this review, the pathological processes and mechanisms of ischemic stroke during perioperative stroke onset were systematically sorted. As a potential treatment strategy for perioperative stroke, the review also summarizes the multifunctional nanodelivery systems based on ischemic stroke, thus providing insight into the nanotherapeutic strategies for perioperative stroke.
Collapse
Affiliation(s)
- Jingyi An
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, China.,Key Laboratories of the Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Ling Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ranran Duan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ke Sun
- Department of Urinary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenxin Lu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiali Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yan Liang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, China.,Key Laboratories of the Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, China.,Key Laboratories of the Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Li Li
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, China.,Key Laboratories of the Ministry of Education, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
39
|
Zheng H, Yuan C, Cai J, Pu W, Wu P, Li C, Li G, Zhang Y, Zhang J, Guo J, Huang D. Early diagnosis of breast cancer lung metastasis by nanoprobe-based luminescence imaging of the pre-metastatic niche. J Nanobiotechnology 2022; 20:134. [PMID: 35292019 PMCID: PMC8922882 DOI: 10.1186/s12951-022-01346-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/02/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Early detection of breast cancer lung metastasis remains highly challenging, due to few metastatic cancer cells at an early stage. Herein we propose a new strategy for early diagnosis of lung metastasis of breast cancer by luminescence imaging of pulmonary neutrophil infiltration via self-illuminating nanoprobes. METHODS Luminescent nanoparticles (LAD NPs) were engineered using a biocompatible, neutrophil-responsive self-illuminating cyclodextrin material and an aggregation-induced emission agent. The chemiluminescence resonance energy transfer (CRET) effect and luminescence properties of LAD NPs were fully characterized. Using mouse peritoneal neutrophils, in vitro luminescence properties of LAD NPs were thoroughly examined. In vivo luminescence imaging and correlation analyses were performed in mice inoculated with 4T1 cancer cells. Moreover, an active targeting nanoprobe was developed by surface decoration of LAD NPs with a neutrophil-targeting peptide, which was also systemically evaluated by in vitro and in vivo studies. RESULTS LAD NPs can generate long-wavelength and persistent luminescence due to the CRET effect. In a mouse model of 4T1 breast cancer lung metastasis, we found desirable correlation between neutrophils and tumor cells in the lungs, demonstrating the effectiveness of early imaging of the pre-metastatic niche by the newly developed LAD NPs. The active targeting nanoprobe showed further enhanced luminescence imaging capability for early detection of pulmonary metastasis. Notably, the targeting nanoprobe-based luminescence imaging strategy remarkably outperformed PET/CT imaging modalities in the examined mouse model. Also, preliminary tests demonstrated good safety of LAD NPs. CONCLUSIONS The neutrophil-targeting imaging strategy based on newly developed luminescence nanoparticles can serve as a promising modality for early diagnosis of lung metastasis of breast cancers.
Collapse
Affiliation(s)
- Hanwen Zheng
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
- Department of Pharmaceutical Analysis, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Chunsen Yuan
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Jiajun Cai
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Wendan Pu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Peng Wu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
- College of Pharmacy and Medical Technology, Hanzhong Vocational and Technical College, Hanzhong, 723000, Shaanxi, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Gang Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Yang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Jiawei Guo
- Department of Pharmaceutical Analysis, College of Pharmacy, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China.
| | - Dingde Huang
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Chongqing, 400038, China.
| |
Collapse
|
40
|
Liu J, Liu Z, Pang Y, Zhou H. The interaction between nanoparticles and immune system: application in the treatment of inflammatory diseases. J Nanobiotechnology 2022; 20:127. [PMID: 35279135 PMCID: PMC8917374 DOI: 10.1186/s12951-022-01343-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/02/2022] [Indexed: 12/24/2022] Open
Abstract
Nanoparticle (NP) is an emerging tool applied in the biomedical field. With combination of different materials and adjustment of their physical and chemical properties, nanoparticles can have diverse effects on the organism and may change the treating paradigm of multiple diseases in the future. More and more results show that nanoparticles can function as immunomodulators and some formulas have been approved for the treatment of inflammation-related diseases. However, our current understanding of the mechanisms that nanoparticles can influence immune responses is still limited, and systemic clinical trials are necessary for the evaluation of their security and long-term effects. This review provides an overview of the recent advances in nanoparticles that can interact with different cellular and molecular components of the immune system and their application in the management of inflammatory diseases, which are caused by abnormal immune reactions. This article focuses on the mechanisms of interaction between nanoparticles and the immune system and tries to provide a reference for the future design of nanotechnology for the treatment of inflammatory diseases.
Collapse
|
41
|
Wu Y, Liu Y, Wang T, Jiang Q, Xu F, Liu Z. Living Cell for Drug Delivery. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
42
|
Advanced drug delivery system against ischemic stroke. J Control Release 2022; 344:173-201. [PMID: 35248645 DOI: 10.1016/j.jconrel.2022.02.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 02/06/2023]
|
43
|
Tang L, He S, Yin Y, Li J, Xiao Q, Wang R, Gao L, Wang W. Combining nanotechnology with the multifunctional roles of neutrophils against cancer and inflammatory disease. NANOSCALE 2022; 14:1621-1645. [PMID: 35079756 DOI: 10.1039/d1nr07725b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Neutrophils, the most abundant leukocytes in humans, play a crucial role in acute inflammation during infection and tumorigenesis. Neutrophils are the major types of cells recruited to the inflammation sites induced by pathogens, exhibiting great homing ability towards inflammatory disorders and tumor sites. Therefore, a neutrophil-based drug delivery system (NDDS) has become a promising platform for anti-cancer and anti-inflammatory treatment. Recent decades have witnessed the huge progress of applying nanomaterials in drug delivery. Nanomaterials are regarded as innovative components to enrich the field of neutrophil-based therapies due to their unique physiochemical characteristics. In this review, the latest advancement of combining diverse nanomaterials with an NDDS for cancer and inflammatory disease treatment will be summarized. It is discussed how nanomaterials empower the therapeutic area of an NDDS and how an NDDS circumvents the limitations of nanomaterials. Moreover, based on the finding that neutrophils are closely involved in the progression of cancer and inflammatory diseases, emerging therapeutic strategies that target neutrophils will be outlined. Finally, as neutrophils were demonstrated to play a central role in the immunopathology of COVID-19, which causes necroinflammation that is responsible for the cytokine storm and sepsis during coronavirus infections, novel therapeutic approaches that anchor neutrophils against the pathological consequences related to COVID-19 will be highlighted as well.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Shun He
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Jing Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Qiaqia Xiao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Ruotong Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Lijun Gao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| |
Collapse
|
44
|
Cardiovascular Nanotechnology. Nanomedicine (Lond) 2022. [DOI: 10.1007/978-981-13-9374-7_12-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
45
|
Li YJ, Wu JY, Liu J, Qiu X, Xu W, Tang T, Xiang DX. From blood to brain: blood cell-based biomimetic drug delivery systems. Drug Deliv 2021; 28:1214-1225. [PMID: 34142628 PMCID: PMC8259840 DOI: 10.1080/10717544.2021.1937384] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 01/04/2023] Open
Abstract
Brain drug delivery remains a major difficulty for several challenges including the blood-brain barrier, lesion spot targeting, and stability during circulation. Blood cells including erythrocytes, platelets, and various subpopulations of leukocytes have distinct features such as long-circulation, natural targeting, and chemotaxis. The development of biomimetic drug delivery systems based on blood cells for brain drug delivery is growing fast by using living cells, membrane coating nanotechnology, or cell membrane-derived nanovesicles. Blood cell-based vehicles are superior delivery systems for their engineering feasibility and versatile delivery ability of chemicals, proteins, and all kinds of nanoparticles. Here, we focus on advances of blood cell-based biomimetic carriers for from blood to brain drug delivery and discuss their translational challenges in the future.
Collapse
Affiliation(s)
- Yong-Jiang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jun-Yong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jihua Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiaohan Qiu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenjie Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Da-Xiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
46
|
Yuan J, Li L, Yang Q, Ran H, Wang J, Hu K, Pu W, Huang J, Wen L, Zhou L, Jiang Y, Xiong X, Zhang J, Zhou Z. Targeted Treatment of Ischemic Stroke by Bioactive Nanoparticle-Derived Reactive Oxygen Species Responsive and Inflammation-Resolving Nanotherapies. ACS NANO 2021; 15:16076-16094. [PMID: 34606239 DOI: 10.1021/acsnano.1c04753] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Stroke is a primary cause of death and disability worldwide, while effective and safe drugs remain to be developed for its clinical treatment. Herein, we report bioactive nanoparticle-derived multifunctional nanotherapies for ischemic stroke, which are engineered from a pharmacologically active oligosaccharide material (termed as TPCD) prepared by covalently conjugating a radical-scavenging compound (Tempol) and a hydrogen-peroxide-eliminating moiety of phenylboronic acid pinacol ester (PBAP) on β-cyclodextrin. Of note, combined functional moieties of Tempol and PBAP on β-cyclodextrin contribute to antioxidative and anti-inflammatory activities of TPCD. Cellularly, TPCD nanoparticles (i.e., TPCD NPs) reduced oxygen-glucose deprivation-induced overproduction of oxidative mediators, increased antioxidant enzyme expression, and suppressed microglial-mediated inflammation, thereby inhibiting neuronal apoptosis. After intravenous (i.v.) delivery, TPCD NPs could efficiently accumulate at the cerebral ischemic injury site of mice with middle cerebral artery occlusion (MCAO), showing considerable distribution in cells relevant to the pathogenesis of stroke. Therapeutically, TPCD NPs significantly decreased infarct volume and accelerated recovery of neurological function in MCAO mice. Mechanistically, efficacy of TPCD NPs is achieved by its antioxidative, anti-inflammatory, and antiapoptotic effects. Furthermore, TPCD NPs can function as a reactive oxygen species labile nanovehicle to efficiently load and triggerably release an inflammation-resolving peptide Ac2-26, giving rise to an inflammation-resolving nanotherapy (i.e., ATPCD NP). Compared to TPCD NP, ATPCD NP demonstrated notably enhanced in vivo efficacies, largely resulting from its additional inflammation-resolving activity. Consequently, TPCD NP-derived nanomedicines can be further developed as promising targeted therapies for stroke and other inflammation-associated cerebrovascular diseases.
Collapse
Affiliation(s)
- Jichao Yuan
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lanlan Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qinghua Yang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hong Ran
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jie Wang
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kaiyao Hu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wendan Pu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jialu Huang
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Wen
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Linke Zhou
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Jiang
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhenhua Zhou
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
47
|
Wang Y, Wang Y, Li S, Cui Y, Liang X, Shan J, Gu W, Qiu J, Li Y, Wang G. Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke. J Nanobiotechnology 2021; 19:331. [PMID: 34674712 PMCID: PMC8529766 DOI: 10.1186/s12951-021-01067-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/29/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Ischemic stroke is an acute and severe neurological disease, and reperfusion is an effective way to reverse brain damage after stroke. However, reperfusion causes secondary tissue damage induced by inflammatory responses, called ischemia/reperfusion (I/R) injury. Current therapeutic strategies that control inflammation to treat I/R are less than satisfactory. RESULTS We report a kind of shield and sword nano-soldier functionalized nanoparticles (monocyte membranes-coated rapamycin nanoparticles, McM/RNPs) that can reduce inflammation and relieve I/R injury by blocking monocyte infiltration and inhibiting microglia proliferation. The fabricated McM/RNPs can actively target and bind to inflammatory endothelial cells, which inhibit the adhesion of monocytes to the endothelium, thus acting as a shield. Subsequently, McM/RNPs can penetrate the endothelium to reach the injury site, similar to a sword, and release the RAP drug to inhibit the proliferation of inflammatory cells. In a rat I/R injury model, McM/RNPs exhibited improved active homing to I/R injury areas and greatly ameliorated neuroscores and infarct volume. Importantly, in vivo animal studies revealed good safety for McM/RNPs treatment. CONCLUSION The results demonstrated that the developed McM/RNPs may serve as an effective and safe nanovehicles for I/R injury therapy.
Collapse
Affiliation(s)
- Yanyun Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Shuyu Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yuliang Cui
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Xiping Liang
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Juanjuan Shan
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Wei Gu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| | - Yiliang Li
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, Guangdong, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
48
|
Wang F, Hou W, Xiao C, Hao Y, Su N, Deng Y, Wang J, Yu L, Xie JM, Xiong JW, Luo Y. Endothelial cell membrane-based biosurface for targeted delivery to acute injury: analysis of leukocyte-mediated nanoparticle transportation. NANOSCALE 2021; 13:14636-14643. [PMID: 34558568 DOI: 10.1039/d1nr04181a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Mimicking and leveraging biological structures and materials provide important approaches to develop functional vehicles for drug delivery. Taking advantage of the affinity and adhesion between the activated endothelial cells and innate immune cells during inflammatory responses, hybrid polyester nanoparticles coated with endothelial cell membranes (EM-P) containing adhesion molecules were fabricated and their capability as vehicles to travel to the acute injury sites through leukocyte-mediated processes was investigated. The in vivo studies and quantitative analyses performed through the lung-inflammation mouse models demonstrated that the EM-Ps preferentially interacted with the neutrophils and monocytes in the circulation and the cellular membrane-based biosurface improved the nanoparticle transportation to the inflamed lung possibly via the motility of neutrophils. Utilizing the transgenic zebrafish model, the leukocyte-mediated transportation and biodistribution of EM-Ps were further visualized in real time at the whole-organism level. Endothelial membranes provided a new biosurface for developing biomimetic vehicles to allow the immune cell-mediated transportation and may enable advanced systems for active and highly efficient drug delivery.
Collapse
Affiliation(s)
- Fang Wang
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Wenda Hou
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Chenglu Xiao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China 100871
| | - Yaoyao Hao
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Ni Su
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Yu Deng
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Jieting Wang
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Luying Yu
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Jing-Ming Xie
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Jing-Wei Xiong
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China 100871
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America 02155
| |
Collapse
|
49
|
He W, Zhang Z, Sha X. Nanoparticles-mediated emerging approaches for effective treatment of ischemic stroke. Biomaterials 2021; 277:121111. [PMID: 34488117 DOI: 10.1016/j.biomaterials.2021.121111] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/20/2022]
Abstract
Ischemic stroke leads to high disability and mortality. The limited delivery efficiency of most therapeutic substances is a major challenge for effective treatment of ischemic stroke. Inspired by the prominent merit of nanoscale particles in brain targeting and blood-brain barrier (BBB) penetration, various functional nanoparticles have been designed as promising drug delivery platforms that are expected to improve the therapeutic effect of ischemic stroke. Based on the complex pathological mechanisms of ischemic stroke, this review outline and summarize the rationally designed nanoparticles-mediated emerging approaches for effective treatment of ischemic stroke, including recanalization therapy, neuroprotection therapy, and combination therapy. On this bases, the potentials and challenges of nanoparticles in the treatment of ischemic stroke are revealed, and new thoughts and perspectives are proposed for the design of feasible nanoparticles for effective treatment of ischemic stroke.
Collapse
Affiliation(s)
- Wenxiu He
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xianyi Sha
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China; The Institutes of Integrative Medicine of Fudan University, 120 Urumqi Middle Road, Shanghai, 200040, China.
| |
Collapse
|
50
|
Han L, Jiang C. Evolution of blood-brain barrier in brain diseases and related systemic nanoscale brain-targeting drug delivery strategies. Acta Pharm Sin B 2021; 11:2306-2325. [PMID: 34522589 PMCID: PMC8424230 DOI: 10.1016/j.apsb.2020.11.023] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/30/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Blood–brain barrier (BBB) strictly controls matter exchange between blood and brain, and severely limits brain penetration of systemically administered drugs, resulting in ineffective drug therapy of brain diseases. However, during the onset and progression of brain diseases, BBB alterations evolve inevitably. In this review, we focus on nanoscale brain-targeting drug delivery strategies designed based on BBB evolutions and related applications in various brain diseases including Alzheimer's disease, Parkinson's disease, epilepsy, stroke, traumatic brain injury and brain tumor. The advances on optimization of small molecules for BBB crossing and non-systemic administration routes (e.g., intranasal treatment) for BBB bypassing are not included in this review.
Collapse
Key Words
- AD, Alzheimer's disease
- AMT, alpha-methyl-l-tryptophan
- Aβ, amyloid beta
- BACE1, β-secretase 1
- BBB, blood–brain barrier
- BDNF, brain derived neurotrophic factor
- BTB, blood–brain tumor barrier
- Blood–brain barrier
- Brain diseases
- Brain-targeting
- CMT, carrier-mediated transportation
- DTPA-Gd, Gd-diethyltriaminepentaacetic acid
- Drug delivery systems
- EPR, enhanced permeability and retention
- GLUT1, glucose transporter-1
- Gd, gadolinium
- ICAM-1, intercellular adhesion molecule-1
- KATP, ATP-sensitive potassium channels
- KCa, calcium-dependent potassium channels
- LAT1, L-type amino acid transporter 1
- LDL, low density lipoprotein
- LDLR, LDL receptor
- LFA-1, lymphocyte function associated antigen-1
- LRP1, LDLR-related protein 1
- MFSD2A, major facilitator superfamily domain-containing protein 2a
- MMP9, metalloproteinase-9
- MRI, magnetic resonance imaging
- NPs, nanoparticles
- Nanoparticles
- P-gp, P-glycoprotein
- PD, Parkinson's disease
- PEG, polyethyleneglycol
- PEG-PLGA, polyethyleneglycol-poly(lactic-co-glycolic acid)
- PLGA, poly(lactic-co-glycolic acid)
- PSMA, prostate-specific membrane antigen
- RAGE, receptor for advanced glycosylation end products
- RBC, red blood cell
- RMT, receptor-mediated transcytosis
- ROS, reactive oxygen species
- TBI, traumatic brain injury
- TJ, tight junction
- TfR, transferrin receptor
- VEGF, vascular endothelial growth factor
- ZO1, zona occludens 1
- siRNA, short interfering RNA
- tPA, tissue plasminogen activator
Collapse
Affiliation(s)
- Liang Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
- Corresponding author. Tel./fax: +86 512 65882089.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 200032, China
| |
Collapse
|